Cancers 16 02796 v2
Cancers 16 02796 v2
Cancers 16 02796 v2
Review
Exploring Predictive and Prognostic Biomarkers in Colorectal
Cancer: A Comprehensive Review
Karam Ashouri 1,† , Alexandra Wong 1,† , Pooja Mittal 1 , Lesly Torres-Gonzalez 1 , Jae Ho Lo 1 , Shivani Soni 1 ,
Sandra Algaze 1 , Taline Khoukaz 1 , Wu Zhang 1 , Yan Yang 1 , Joshua Millstein 2 , Heinz-Josef Lenz 1, *,‡
and Francesca Battaglin 1, *,‡
1 Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine,
University of Southern California, Los Angeles, CA 90089, USA; [email protected] (K.A.);
[email protected] (A.W.)
2 Department of Population and Public Health Sciences, Norris Comprehensive Cancer Center,
Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
* Correspondence: [email protected] (H.-J.L.); [email protected] (F.B.);
Tel.: +1-(323)-865-3967 (H.-J.L.); +1-(323)-865-3930 (F.B.)
† These authors contributed equally to this work as co-first authors.
‡ These authors contributed equally to this work as co-last authors.
Simple Summary: Colorectal cancer is a major health concern globally, and finding ways to improve
treatment outcomes is crucial. This review explores the role of biomarkers—biological indicators that
can predict how a patient will respond to treatment or indicate the likely course of the disease—in
managing colorectal cancer. By examining both well-established and emerging biomarkers, we hope
to provide a clearer understanding of how these markers can guide personalized treatment plans. The
findings from this research could help doctors make more informed decisions, ultimately improving
patient care and outcomes in colorectal cancer.
Abstract: Colorectal cancer (CRC) remains the second leading cause of cancer-related mortality
worldwide. While immune checkpoint inhibitors have significantly improved patient outcomes,
Citation: Ashouri, K.; Wong, A.; their effectiveness is mostly limited to tumors with microsatellite instability (MSI-H/dMMR) or
Mittal, P.; Torres-Gonzalez, L.; Lo, J.H.; an increased tumor mutational burden, which comprise 10% of cases. Advancing personalized
Soni, S.; Algaze, S.; Khoukaz, T.;
medicine in CRC hinges on identifying predictive biomarkers to guide treatment decisions. This
Zhang, W.; Yang, Y.; et al. Exploring
comprehensive review examines established tissue markers such as KRAS and HER2, highlighting
Predictive and Prognostic Biomarkers
their roles in resistance to anti-EGFR agents and discussing advances in targeted therapies for these
in Colorectal Cancer: A
markers. Additionally, this review summarizes encouraging data on promising therapeutic targets
Comprehensive Review. Cancers 2024,
16, 2796. https://doi.org/10.3390/
and highlights the clinical utility of liquid biopsies. By synthesizing current evidence and identifying
cancers16162796 knowledge gaps, this review provides clinicians and researchers with a contemporary understanding
of the biomarker landscape in CRC. Finally, the review examines future directions and challenges
Academic Editor: Tianhui Hu
in translating promising biomarkers into clinical practice, with the goal of enhancing personalized
Received: 28 June 2024 medicine approaches for colorectal cancer patients.
Revised: 4 August 2024
Accepted: 7 August 2024 Keywords: colorectal cancer; gene expression profiling; molecular targeted therapy; tumor biomarkers
Published: 8 August 2024
1. Introduction
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
Colorectal cancer (CRC) is the third most common cancer and the second leading cause
This article is an open access article
of cancer-related deaths worldwide [1]. Approximately 20% of patients have metastatic
distributed under the terms and disease upon presentation, and the 5-year survival rate for these patients remains less than
conditions of the Creative Commons 20%, highlighting the need to expand the therapeutic armamentarium in this setting [2].
Attribution (CC BY) license (https:// Over the last two decades, the treatment landscape for metastatic CRC (mCRC) has
creativecommons.org/licenses/by/ significantly evolved. The standard of treatment was 5-fluorouracil (5-FU) combined with
4.0/). leucovorin until it was found that combining 5-FU with oxaliplatin or irinotecan improved
median overall survival (OS) to 20 months [3]. The advent of immunotherapy has also led to
substantial therapeutic benefits, with a 4-year OS > 70% in mCRC positive for microsatellite
instability or mismatch repair deficiency (MSI-H/dMMR) [4]. However, this biomarker
applies to 4% of all mCRC cases, leaving most patients reliant on traditional chemotherapy
and incremental gains from new biologic agents.
The identification of predictive and prognostic biomarkers with immunohistochem-
istry (IHC) or next-generation sequencing (NGS) is crucial for advancing personalized
medicine in CRC. Predictive biomarkers help determine the likelihood of the response to
specific therapies, while prognostic biomarkers provide information on the overall dis-
ease outcome, regardless of treatment. Notably, CRC primary tumors and metastatic sites
harbor a high concordance (>90%) in their mutational profile for actionable genes [5,6].
Integrating these genetic markers in the four distinct consensus molecular subtypes (CMSs)
has enriched our understanding of the clinical and biological characteristics of CRC [7].
Despite the progress in molecular diagnostics, challenges remain in translating these
biomarkers into clinical practice. The heterogeneity of CRC, both morphologically and
molecularly, complicates the identification of universally applicable biomarkers. The
clinical relevance of many emerging biomarkers is still under investigation, necessitating
further research and validation in larger patient cohorts. This review aims to provide a
comprehensive overview of key established, promising, and potential biomarkers in CRC.
2. Established Biomarkers
2.1. RAS Status
Proteins of the RAS family are GTPases involved in the RAS/RAF/MEK/ERK path-
way, playing a crucial role in cell division and proliferation. RAS mutations are prevalent
across various cancers, making them one of the most common oncogenes. In CRC, RAS
mutations are found in approximately 55% of cases, with KRAS mutations occurring in
about 50%, NRAS in 4%, and HRAS in less than 1% [8].
Most KRAS mutations involve glycine substitutions at positions 12 or 13, leading
to constitutive activation of the GTPase side chain. The most common KRAS mutations
in CRC are KRAS G12D (36%), KRAS G12V (21.8%), and KRAS G13D (18.8%), while
KRAS G12C mutations are found in 3–4% of CRC cases (Figure 1) [9,10]. KRAS muta-
tions are more prevalent in left-sided tumors than right-sided tumors and similarly more
common in microsatellite stable (MSS) CRC relative to MSI-H/dMMR CRC. In frontline
chemotherapy–treated mCRC, the KRAS wildtype status conferred an improved OS relative
to mutated tumors (20.9 months vs. 16.9 months, HR = 0.71, 95% CI 0.57, 0.88, HORIZON II,
NCT00399035) [11]. The prognostic utility of KRAS mutations extends to both young-onset
and late-onset CRC, revealing worse cause-specific survival relative to KRAS wildtype
tumors in both age groups [12]. Among specific KRAS mutations, G12C demonstrated the
worst prognosis relative to wildtype tumors (4.3 months vs. 23.3 months, p < 0.001) [13].
RAS mutations result in constitutive activation downstream of EGFR blockade, ren-
dering epidermal growth factor receptor (EGFR) inhibitors ineffective. Multiple studies, in-
cluding the phase 3 CRYSTAL and PRIME trials, have demonstrated that KRAS/NRAS mu-
tations predict a poor response to combination chemotherapy with EGFR inhibitors [14,15].
Consequently, testing for KRAS/NRAS mutations is recommended by the National Com-
prehensive Cancer Network (NCCN) for all patients with mCRC, because those with these
mutations should not receive panitumumab or cetuximab in combination with chemother-
apy or as monotherapy [16]. Within 12 months of treatment, most CRCs acquire resistance
to anti-EGFR therapies, frequently (~50%) due to secondary KRAS alterations [17,18]. This
underscores the importance of checking for these mutations during anti-EGFR therapy.
ers 2024, 16, x FOR PEER REVIEW 3 o
Cancers 2024, 16, 2796 3 of 25
FigureFigure
1. Current predictive biomarkers for metastatic colorectal cancer (CRC). EGFRi = EGFR i
1. Current predictive biomarkers for metastatic colorectal cancer (CRC). EGFRi = EGFR
bition;inhibition;
VEGFi =VEGFi
VEGF= inhibition; G12Ci
VEGF inhibition; = KRAS
G12Ci = KRASG12C
G12C inhibition; BRAFi
inhibition; BRAFi = BRAF
= BRAF inhibition; I
inhibition;
Immune checkpoint inhibition; HER2i = HER2 inhibition; CRC = colorectal cancer. (Created
ICI = Immune checkpoint inhibition; HER2i = HER2 inhibition; CRC = colorectal cancer. (Created in in
render.com).
Biorender.com).
mors [19]. Updated results from the RAS wildtype cohort in the FIRE-3 trial indicated
improved OS with the FOLFIRI plus cetuximab group compared to the FOLFIRI plus beva-
cizumab group (33.1 months vs. 25.0 months; HR = 0.70; p = 0.0059) [20]. The cetuximab
group also demonstrated a more frequent early tumor shrinkage and a greater median
depth of response [20].
Recent advances have led to the development of oral KRAS G12C small molecule
inhibitors. Initial studies of sotorasib and adagrasib monotherapy showed modest effects
in chemo-refractory mCRC, and combining them with EGFR inhibitors appears to en-
hance their efficacy and overcome resistance mechanisms [21]. The KRYSTAL-1 trial, a
phase I/II study examining adagrasib with or without cetuximab, showed a numerically
higher median duration of response and response rates with the addition of cetuximab [22]
(NCT03785249). Soon after, adagrasib was awarded accelerated Food and Drug Administra-
tion (FDA) approval as a second-line agent in combination with cetuximab for KRAS G12C–
mutated CRC. The phase 3 CodeBreak 300 trial evaluates sotorasib in chemo-refractory
mCRC at two doses (240 mg and 960 mg) in combination with panitumumab compared
to a standard of care (SOC) group (Lonsurf or regorafenib) [23]. Early data demonstrate
superior progression free survival (PFS) in both sotorasib groups relative to SOC (960 mg:
5.6 months vs. 240 mg: 3.9 months vs. SOC: 2.2 months) (NCT05198934) [23]. Another
KRAS G12C inhibitor, divarasib, is being studied in a Phase 1 trial as monotherapy and
in combination with cetuximab (NCT04449874) (Table 1). Additional phase 3 studies will
examine adagrasib combined with cetuximab compared to chemotherapy in the second-line
setting (NCT04793958) and sotorasib in combination with panitumumab and chemotherapy
in treatment-naive patients (NCT06252649). Preclinical data suggest that SHP2-inhibitors
augment KRAS G12C inhibitors by improving engagement and downregulating resis-
tance mechanisms, leading to clinical trials evaluating this combination (NCT04330664,
NCT04916236, NCT05288205, NCT04699188, NCT04449874) (Table 1) [24]. In preclinical
CRC models, KRAS mutations increased the expression of amino acid transporters via
the hippo pathway effector YAP1, resulting in mTOR activation and subsequent CRC cell
proliferation [25]. Mechanistic studies in lung cancer indicate that the hippo pathway is
implicated in KRAS G12C resistance, highlighting its therapeutic potential in augmenting
KRAS inhibitors [26]. G12D is the most prevalent KRAS alteration in CRC, with several
small molecular inhibitors (MRTX1133 and RMC-9805) undergoing early phase evaluation
(NCT05737706, NCT06040541) (Table 1). MRTX1133 (G12D inhibitor) has shown synergism
alongside 5-FU in preclinical models, with similar effects in KRAS G12V–mutated cell
lines [27]. Alternative strategies targeting specific KRAS mutations utilize T cell receptors
(TCRs), which recognize intracellular antigens presented by human leukocyte antigen
(HLA) molecules to trigger an immune response [28]. Several clinical trials utilize TCRs
in KRAS G12V (NCT06105021, NCT06043713) and G12D (NCT03948763) CRC (Table 1).
In addition to the mutation selective inhibitors, clinical trials are evaluating pan-KRAS
inhibitors (NCT04975265) (Table 1). RMC-6236 is a pan-RAS inhibitor that targets the full
range of RAS alterations (KRAS, HRAS, NRAS) with activity against mutant and wildtype
RAS variants and is undergoing Phase 1 studies (NCT05379985) (Table 1) [29].
Cancers 2024, 16, 2796 5 of 25
Table 1. Clinical trials evaluating therapeutic potential in KRAS mutations and promising biomarkers.
mCRC Sotorasib (G12C inhibitor) + panitumumab + NCT06252649 (Phase 3) Not yet recruiting
FOLFIRI
KRAS G12C mutation
mCRC Divarasib (G12C inhibitor) + cetuximab NCT04449874 (Phase 1) Active
Glecirasib (G12C inhibitor) + JAB-3312 Active
Advanced solid tumors (SHP2 inhibitor) NCT05288205 (Phase 1/2)
Advanced solid tumors Opnurasib (G12C inhibitor) NCT04699188 (Phase 1/2) Active
Advanced solid tumors Divarasib (G12C inhibitor) NCT04449874 (Phase 1) Active
KRAS KRAS mutated (except Vociprotafib
mCRC NCT04916236 (Phase 1) Active
G13 mutations) (SHP2 inhibitor) + Temuterkib (ERK ½ inhibitor)
Adagrasib (G12C inhibitor) + Batoprotafib
mCRC NCT04330664 (Phase 1/2) Active
(SHP2 inhibitor)
KRAS G12D mutation Advanced solid tumors MRTX1133 (G12D inhibitor) NCT05737706 (Phase 1/2) Active
Advanced solid tumors RMC-9805 (G12D inhibitor) NCT06040541 (Phase 1) Active
Advanced solid tumors AFNT-211 (G12V TCR) NCT06105021 (Phase 1/2) Active
KRAS G12V mutation
Advanced solid tumors FHA11 (G12V TCR) NCT06043713 (Phase 1) Active
KRAS G12D, G12V, G13D or
G12C Non-MSI-H/dMMR mCRC mRNA-5671/V941 NCT03948763 (Phase 1) Completed (2022)
RAS mutation Advanced solid tumors RMC-6236 (RASmulti inhibitor) NCT05379985 (Phase 1) Active
CEACAM5 Advanced solid tumors M9140 (CEACAM5 ADC) NCT05464030 (Phase 1) Active
Obinutuzumab (CD20 mAb) + Cibisatamab
High CEACAM5 Expression Previously treated MSS mCRC (CEA-CD3 bispecific Ab) + atezolizumab NCT03866239 (Phase 1) Completed (2024)
(PDL1 mAb)
c-MET Advanced solid tumors ABBV-400 (MET Ab) + Bevacizumab NCT05029882 (Phase 1) Active
Previously treated unresectable ABBV-400 (MET Ab) + 5FU + Folinic acid + NCT06107413 (Phase 2) Active
mCRC Bevacizumab
PIK3CA mutation Advanced solid cancers including Serabelisib (PI3Kα inhibitor) NCT05300048 (Phase 1b) Active
PIK3CA CRC
PIK3CA mutation mCRC Alpelisib (PI3Kα inhibitor) + capecitabine NCT04753203 (Phase 1b/2) Active
Cancers 2024, 16, 2796 6 of 25
Table 1. Cont.
KRAS or NRAS Mutation mCRC Onvansertib (PLK1 inhibitor) + Bevacizumab + NCT05593328 (Phase 2) Active
FOLFIRI
CRC = colorectal cancer; mCRC = metastatic colorectal cancer; MSS = microsatellite stable; CPM = composite PD-L1/Mucin; CPS = combined positive score.
Cancers 2024, 16, 2796 7 of 25
2.3. HER2
HER2, encoded by ERBB2, is an EGFR tyrosine kinase family member. Its overex-
pression and/or amplification have been reported in various solid tumors, particularly
breast cancer, where HER2-targeted therapies have been highly successful. The NCCN
Guidelines now recommend HER2 testing for all mCRC patients, present in 2–6% of cases
given therapeutic implications with anti-EGFR and HER2 directed therapy (Figure 1).
HER2 is measured by IHC, fluorescence in situ hybridization (FISH), or NGS. HER2 over-
expression is indicated by an IHC score of 3+, while an IHC score of 2+ is equivocal and
should prompt FISH testing. A FISH ratio ≥2 confirms HER2 amplification [54]. HER2 am-
plification is more common in left-sided tumors and is more prevalent in RAS/BRAF
wildtype tumors [55,56]. Studies have shown that HER2 overexpression may confer re-
Cancers 2024, 16, 2796 8 of 25
sistance to EGFR inhibition (Figure 1) [55,57,58]. On the other hand, a recent analysis
from the CALGB/SWOG 80405 trial found that high HER2 expressing mCRC (tumor gene
expression above the median) had a longer PFS and OS than tumors with lower HER2
expression [59]. In patients with lower HER2 expression, treatment with cetuximab was
associated with a worse PFS and OS compared to bevacizumab. The poor prognostic
implication of decreased HER2 expression was confirmed in RAS wildtype mCRC based
on the MONSTAR-SCREEN-2 cohort [60]. These findings suggest that HER2 expression
is prognostic and may contribute to selecting anti-EGFR versus anti-VEGF treatment in
the first-line metastatic setting [59]. However, other studies have reported mixed findings
regarding the prognostic role of HER2, and further prospective validation is needed [61].
HER2 is an established therapeutic target in CRC. In the second-line setting for mCRC,
RAS/BRAF wildtype patients with HER2 overexpression/amplification who have not
received prior HER2 inhibitors can be treated with trastuzumab in combination with
pertuzumab, lapatinib, or tucatinib. The HERACLES trial, a phase 2 study of heavily
pre-treated patients with the KRAS wildtype, HER2+ mCRC, demonstrated the efficacy
of dual HER2 blockade using trastuzumab and lapatinib, with an objective response rate
(ORR) of 30% [56]. The MyPathway basket study of trastuzumab and pertuzumab in
treatment-refractory CRC reported an ORR of 38% [62]. The combination of trastuzumab
and tucatinib was the first HER2-directed treatment approved by the FDA for chemo-
refractory mCRC, based on the MOUNTAINEER trial (ORR 38%) [63]. The ongoing phase
3 trial, MOUNTAINEER-03 (NCT05253651), is investigating tucatinib and trastuzumab
in combination with chemotherapy in the first-line setting compared to standard care in
HER2+ patients. In the DESTINY-CRC01 phase 2 trial of patients with HER2+ mCRC who
had received at least two prior lines of therapy, fam-trastuzumab deruxtecan was associated
with an ORR of 45% [64]. Notably, fam-trastuzumab deruxtecan is an option for patients
who received prior HER2-directed therapy; however, the cohort included only patients
with HER2 IHC 3+, and patients must be monitored closely for the potential serious side
effect of interstitial lung disease [64]. Future studies will help elucidate ideal sequencing
strategies for targeted therapy in this group of patients.
2.4. MSI-H
Microsatellites are short tandem repeated sequences spread throughout DNA. Mi-
crosatellite instability is caused by defects in the mismatch repair system, most commonly
due to somatic mutations of MLH1, MSH2, MSH6, PMS2, and EPCAM, which lead to
defects during DNA replication [65,66]. The prevalence of MSI-H/dMMR decreases with
the advancing CRC stage as follows: stage II (20%), then stage III (12%), and stage IV
(4%) (Figure 1) [67]. Knowledge of the MSI-H/dMMR status is critical in managing CRC
for several reasons. Patients with MSI-H/dMMR tumors must be screened for germline
mutations in the mismatch repair genes, which leads to Lynch Syndrome, altering the
treatment of patients and relatives [68]. With the introduction of immune checkpoint
inhibitors (ICI), MSI-H/dMMR status has offered profound survival benefits and cures
for some CRCs [69]. The high neoantigen load of MSI-H/dMMR cancer elicits a strong
antitumor immune response, further augmented with ICIs (Figure 1) [70]. Additionally,
MSI-H/dMMR CRCs have a favorable tumor microenvironment (TME) with increased
tumor-infiltrating lymphocytes and decreased immunosuppressive cells like regulatory T
cells (Tregs) and myeloid-derived suppressor cells (MDSCs) [71,72]. ICIs further enhance
the activity of these preexisting tumor infiltrating lymphocytes (TILs) to target the tumor.
The phase II KEYNOTE-016 trial was the first to demonstrate superior ORR (40%) in
MSI-H/dMMR mCRC relative to MSS (0%) with pembrolizumab [73]. Pembrolizumab’s
superior antitumor activity was confirmed in the KEYNOTE-164 trial, demonstrating a
median PFS and OS of 4.1 and 47.0 months, respectively, in pretreated CRC (chemotherapy
arm PFS: 2.3 months, OS: 31.4 months) [69,74]. The 50% increase in OS relative to the SOC
and 30% PFS plateau at 5 years, all with fewer adverse events in the experimental arm, were
unheard of in previous CRC trials. A similar efficacy was demonstrated with nivolumab
Cancers 2024, 16, 2796 9 of 25
(phase II Checkmate 142 trial), awarding both agents FDA approval for second-line use in
2017 [75]. Phase III KEYNOTE-177 demonstrated that first-line pembrolizumab (200 mg
q3w for up to 2 years) was superior to standard chemotherapy (mFOLFOX6 or FOLFIRI
q2w ± bevacizumab or cetuximab) in MSI-H/dMMR mCRCs, confirming a longer median
PFS (16.5 months vs. 8.2 months, HR 0.60, p = 0.0002) [76]. The effect of nivolumab was
augmented with the addition of anti-CTLA4 (ipilimumab) therapy (24-month PFS: 74%
and OS: 79%) [77]. Phase III Checkmate-8HW confirmed the superior PFS of nivolumab
plus ipilimumab compared to chemotherapy (not reached (NR) vs. 5.9 months; HR 0.21,
p < 0.0001) after 31.5 months of median follow-up in the first-line setting [4].
Despite the clinical benefit, nearly 30% of MSI-H/dMMR tumors are resistant to
ICI [78]. Preclinical data indicate that inhibition of VEGF may promote an immune permis-
sive TME [79,80]. Following encouraging safety and efficacy data, atezolizumab plus beva-
cizumab plus FOLFOX is being evaluated as a first-line regimen in MSI-H/dMMR mCRC
(NCT02997228) [81]. Additional studies combine VEGF inhibitors with pembrolizumab or
nivolumab as second-line agents (NCT05035381).
NRTK fusion is comparably less (commonly <5%) in common cancers, with the frequency
being 0.7–1.5% in CRC (Figure 1) [89].
A recent report by Wang et al. (2022) documented that frequent coalterations in APC
and TP53 genes were found with NTRK fusions, while alterations in RAS/BRAF were
mutually exclusive. NTRK fusion-positive tumors showed a high TMB, MSI-H/dMMR,
and enrichment in POLE/POLD1 mutations compared to molecularly unstratified CRC
tumors. Typically, the NTRK fusion proteins contain the kinase domain of NTRK juxta-
posed with a different gene. In several cases, this leads to the constitutive activation of
downstream signaling pathways such as MAPK and PI3K. NTRK fusions are clinically
targetable mutations, and two first-generation NTRK tyrosine kinase inhibitors (TKIs),
larotrectinib and entrectinib, have been approved by the FDA for the treatment of both
adult and pediatric cancers (Figure 1). Second or next-generation NTRK inhibitors, which
could overcome the resistance to first-generation NTRK TKIs, are also in clinical trials,
including LOXO-195 and TPX-0005 [88,90].
Another study documented the impact of NTRK fusions on outcomes in patients with
locally advanced or metastatic solid cancers. The NTRK fusion-positive cohort (which
included 10 different cancer types with the highest number of patients having CRC) showed
a hazard ratio of 1.6 (p = 0.05) and a median OS of 10.2 months compared to the NTRK
fusion-negative cohort (median OS = 10.4 months). This study suggested that NTRK
fusions might be a negative prognostic factor; however, further studies are warranted [91].
3. Promising Biomarkers
3.1. PD-L1
PD-L1 is expressed at low levels on antigen-presenting cells and a variety of non-
hematopoietic cells, binding PD-1 on T cells [92]. This interaction inhibits T cell activation
and proliferation, suppressing the immune response against tumor cells and allowing
them to evade immune surveillance [93]. In theory, higher PD-L1 expression in tumor
tissue should confer an improved response to ICI treatment. While PD-L1 is a predictive
biomarker in NSCLC, melanoma, and renal cell cancer, its use in CRC is limited [94,95].
PD-L1 expression is induced by IFN-y and TNF-α, secreted by TILs, particularly CD8+ T
cells [92]. Therefore, PD-L1 expression by IHC is highly dependent on spatial heterogeneity
and sampling. Additionally, the plethora of primary antibodies, staining conditions, and
intertumoral heterogeneity add to the technical and biological discrepancy in PD-L1 expres-
sion, limiting its use as a biomarker [96]. Analysis of nivolumab plus ipilimumab–treated
MSI-H/dMMR CRC showed no difference in ORR utilizing PD-L1 ≥ 1% as a cutoff [97].
Meta-analyses have demonstrated positive tumor cell PD-L1 expression to confer worse
OS, but other studies did not find this association or found the opposite effect [98–100].
Some studies have highlighted an increased PD-1 expression in immune cells and TILs to
confer an improved OS in CRC [100,101]. While the utility of PD-1/PD-L1 as a biomarker
requires further research, additional therapies targeting PD-1, such as balstilimab, are being
investigated in colorectal cancer (NCT05608044).
3.2. PI3K
PIK3CA is a key driver gene that encodes a subunit of phosphatidylinositol 3-kinase
(PI3K), a critical component of the PI3K/AKT/mTOR pathway involved in prolifera-
tion, survival, and angiogenesis [102]. PIK3CA mutations activate the PI3K/AKT/mTOR
pathway downstream of RAS activation, conferring resistance to anti-EGFR therapy in
KRAS-wildtype mCRC, with a more pronounced effect in exon 20 alterations [103–105].
The prognostic implication of PIK3CA mutations remains unclear in CRC. Although some
studies have reported a worse prognostic effect with PIK3CA mutations in mCRC, others
have reported a null association [104,106]. Evaluation of serabelisib (PIK3 inhibitor) in solid
tumor patients with PIK3CA mutations is in a phase 1b trial (NCT05300048) (Table 1).
Cancers 2024, 16, 2796 11 of 25
3.5. TIM3/LAG3
T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) and lymphocyte-
activation gene 3 (LAG3) are immune checkpoint receptors expressed on T cells, macrophages, and
other immune cells, which suppresses antitumor immune responses [122,123]. Within CRC
TME, TIM3 and LAG3 promote immune tolerance by inhibiting T cell activity and favoring
M2-like polarization of macrophages [124,125]. In stage I-III CRC, an increased tumor TIM3
and LAG3 expression correlated with a decreased OS, while an increased expression on
immune cells had the opposite effect [126]. Murine models revealed that the co-blockade
of TIM3 and its ligand (CEACAM1) leads to enhanced antitumor immune responses, im-
proving CRC clearance [127]. Clinical trials are evaluating TSR-022 (anti TIM3 antibody) in
solid tumors, including colorectal cancer (phase 1 NCT02817633). Mouse models demon-
strated that combining LAG3 and PD1 inhibitors overcame tumor resistance to either agent
alone [128]. Similarly, the evaluation of nivolumab with relatlimab (LAG3 monoclonal
antibody) in MSS colorectal cancer patients is underway (phase 2 NCT03642067) (Table 1).
After a combination of favezelimab (Anti-LAG3) and pembrolizumab demonstrated safety
and robust efficacy in the PD-L1 combined positive score (CPS) ≥ 1 MSS mCRC, a phase
3 trial is comparing it to SOC in PD-L1 positive CRC (NCT05064059) (Table 1) [129].
3.6. CEA/CEACAM5
Carcinoembryonic antigen (CEA), a glycoprotein and tumor marker encoded by CEA-
CAM5, is elevated in patients with CRC. Preoperative serum CEA (s-CEA) of > 5 ng/mL
confer worse OS and cancer-specific survival [127,128]. In addition to s-CEA, tissue CEA
(t-CEA) can be assessed using IHC with expression patterns described as apicoluminal
Cancers 2024, 16, 2796 12 of 25
3.7. c-MET
Mesenchymal–epithelial transition factor (c-MET) is a receptor tyrosine kinase encoded
by MET, responsible for cellular proliferation, angiogenesis, and epithelial–mesenchymal
transition (EMT) [130]. Several studies, along with a meta-analysis, noted that an increased
c-MET expression predicts worse OS and DFS [131,132]. MET amplification correlates
with acquired resistance to anti-EGFR therapy in CRC without KRAS mutations [133].
Combining cetuximab with MET inhibitors suppressed MET-induced anti-EGFR resistance
in CRC cell lines [134]. Early data on ABBV-400, a c-MET-targeted ADC linked with
telisotuzumab (topoisomerase 1 inhibitor), demonstrated a robust safety profile and ORR
(61.9%) in CRC (NCT05029882, NCT06107413) (Table 1) [135].
3.8. ARID1A
ARID1A is a subunit of the SWI/SNF complex, which plays a role in regulating
DNA repair. A loss of function of the SWI/SNF complex promotes genomic instability
and tumor progression. In CRC, ARID1A is a known driver gene, and mutations are
associated with right-sided tumors, MSI-H/dMMR, and BRAF mutations [136]. Studies
suggest that ARID1A mutations may be implicated in both intrinsic and acquired resistance
to cetuximab [137]. Patients enrolled in CALGB/SWOG 80405, a randomized phase III
study evaluating the efficacy of chemotherapy plus cetuximab vs. chemotherapy plus
bevacizumab in first-line CRC, who had ARID1A mutations at baseline, had poorer clinical
outcomes with cetuximab compared to bevacizumab. These findings were supported
in patient-derived xenograft models of extended RAS/BRAF wildtype tumors. In post-
treatment cell-free DNA (cfDNA) analysis of these patients, KRAS and ARID1A mutations
were enriched in those who were treated with cetuximab but not bevacizumab. Additional
CRC genomic analysis suggests that ARID1A and EGFR-pathway mutations are mutually
exclusive, consistent with findings in lung cancer. Further exploration is warranted to
delineate if ARID1A mutations identify a group of patients who benefit from MAPK
inhibition, in addition to being a predictive biomarker of a poor response to cetuximab
treatment. Phase 2 trials are evaluating tislelizumab (PD-1 Antibody) plus fruquintinib
(VEGFR 1/2/3 Inhibitor) for ARID1A-mutated MSS mCRC (NCT05690035) (Table 1).
3.9. PLK1
Polo-like Kinase 1 (PLK1) is a serine/threonine protein kinase family that plays a
role in cell cycle progression by regulating progression to mitosis at the G2/M check-
point [138]. It also plays a role in DNA damage response and cell death pathways. PLK1 is
overexpressed in many tumor types and associated with poor prognosis, making it a
promising therapeutic target. Higher levels of PLK-1/phosphorylated-PLK1 were found
in relapsed/mCRC tissues compared to matched primary CRC tissues, suggesting that
it is a marker of poor prognosis and resistance to oxaliplatin-based chemotherapy [139].
Inhibition of PLK1 in CRC cell lines demonstrated that PLK1 inhibitors may attenuate
oxaliplatin resistance [139]. A Phase 1b/2 trial studied onvansertib, an oral highly selective
PLK1 inhibitor, in combination with FOLFIRI and bevacizumab as a second-line treat-
ment for KRAS-mutated mCRC and found that the combination was well-tolerated with
durable responses [140]. A Phase 2 trial is underway to assess its utility combined with
Cancers 2024, 16, 2796 13 of 25
5. Future Directions
NGS has altered the paradigm of genomics research, offering unparalleled capabilities
for analyzing DNA and RNA molecules in a high-throughput and cost-effective manner.
Third-generation sequencing has allowed for longer read lengths, real-time sequencing,
minimal sample preparation, and direct detection of epigenetic modifications, providing
a comprehensive understanding of genomic structure [151]. Using NGS in ctDNA offers
a minimally invasive approach to monitor disease progression and treatment response.
Studying the dynamics of these biomarkers over time and across different treatment
regimens can provide valuable insights into the tumor landscape and inform personalized
treatment strategies [152].
CRC is known to exhibit a high intratumor heterogeneity, with different subpopula-
tions of tumor cells harboring distinct genetic and transcriptomic profiles. Single-cell RNA
sequencing (scRNA-seq) evaluates the heterogeneity at the single-cell level, enabling the
identification of rare cell subpopulations that may play crucial roles in disease progression,
metastasis, and treatment resistance [153]. Additionally, scRNA-seq can delineate the com-
plex interactions between tumor cells and immune cells, potentially identifying biomarkers
related to immunosuppression or immune evasion.
Cancers 2024, 16, 2796 14 of 25
cellular metabolism, limiting these processes and thereby making KRAS-mutant tumors
more susceptible to ferroptosis induction as a potential clinical strategy [191].
As the era of targeted therapies continues to evolve, the development of companion
diagnostics and predictive biomarkers becomes increasingly crucial. Identifying biomarkers
that can accurately predict the response to targeted agents will maximize therapeutic
efficacy while minimizing unnecessary toxicities and healthcare costs.
6. Conclusions
The predictive capacity of MSI-H, RAS, and BRAF has necessitated a molecular-
targeted approach in CRC. The MSI status confers substantial improvements in clinical
outcomes and decreased toxicities through ICI, offering patients long-term survival or near
curative results [69]. BRAF V600E had a negative prognostic value but is now predictive
of the response to combined targeted BRAF/EGFR inhibition, doubling survival over
standard treatments [51]. While the prevalence of these alterations constitutes the minority
of CRCs, they underscore the necessity of identifying predictive biomarkers.
Author Contributions: All authors have made substantial contributions to the review. Conceptual-
ization: K.A., A.W., H.-J.L. and F.B.; writing, review, and/or revision of the manuscript: all authors;
study supervision/guarantor: K.A., A.W., H.-J.L. and F.B. All authors have read and agreed to the
published version of the manuscript.
Funding: This study was partly supported by the National Cancer Institute (grant number P30CA014089)
[to H.-J.L.], the Gloria Borges WunderGlo Foundation, Dhont Family Foundation, Victoria and Philip
Wilson Research Fund, San Pedro Peninsula Cancer Guild, Ming Hsieh Research award, and Daniel
Butler Research Fund. The content is solely the responsibility of the authors and does not necessarily
represent the official views of the National Institutes of Health.
Conflicts of Interest: H.-J.L. reports receiving honoraria from consultant/advisory board membership
for Merck Serono, Bayer, and Genentech. The remaining authors declare that the research was
conducted in the absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Abbreviations
5-FU 5-fluorouracil
ADC antibody-drug conjugate
AI artificial intelligence
AL apicoluminal
BRAFV600E BRAF mutation with valine substitution for glutamic acid at amino acid 600
c-MET mesenchymal–epithelial transition factor
CCR5 C-C motif chemokine receptor 5
CEA carcinoembryonic antigen
cfDNA cell-free DNA
CMS consensus molecular subtype
CRC colorectal cancer
ctDNA circulating tumor DNA
CXCR4 CXC chemokine receptor 4
DC diffuse-cytoplasmic
DFS disease-free survival
DPD dihydropyrimidine dehydrogenase
EGFR epidermal growth factor receptor
EMT epithelia–mesenchymal transition
FDA Food and Drug Administration
FISH fluorescence in situ hybridization
GSH glutathione
HLA human leukocyte antigen
Cancers 2024, 16, 2796 16 of 25
References
1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN
estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [CrossRef]
[PubMed]
2. Biller, L.H.; Schrag, D. Diagnosis and Treatment of Metastatic Colorectal Cancer: A Review. JAMA 2021, 325, 669–685. [CrossRef]
3. Grothey, A.; Sargent, D.; Goldberg, R.M.; Schmoll, H.J. Survival of patients with advanced colorectal cancer improves with the
availability of fluorouracil-leucovorin, irinotecan, and oxaliplatin in the course of treatment. J. Clin. Oncol. 2004, 22, 1209–1214.
[CrossRef] [PubMed]
4. Lenz, H.-J.; Lonardi, S.; Elez, E.; Cutsem, E.V.; Jensen, L.H.; Bennouna, J.; Mendez, G.; Schenker, M.; Fouchardiere, C.D.L.; Limon,
M.L.; et al. Nivolumab (NIVO) plus ipilimumab (IPI) vs chemotherapy (chemo) as first-line (1L) treatment for microsatellite
instability-high/mismatch repair-deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC): Expanded efficacy analysis
from CheckMate 8HW. J. Clin. Oncol. 2024, 42, 3503. [CrossRef]
5. Lee, S.E.; Park, H.Y.; Hwang, D.Y.; Han, H.S. High Concordance of Genomic Profiles between Primary and Metastatic Colorectal
Cancer. Int. J. Mol. Sci. 2021, 22, 5561. [CrossRef] [PubMed]
6. Brannon, A.R.; Vakiani, E.; Sylvester, B.E.; Scott, S.N.; McDermott, G.; Shah, R.H.; Kania, K.; Viale, A.; Oschwald, D.M.; Vacic, V.;
et al. Comparative sequencing analysis reveals high genomic concordance between matched primary and metastatic colorectal
cancer lesions. Genome Biol. 2014, 15, 454. [CrossRef] [PubMed]
7. Guinney, J.; Dienstmann, R.; Wang, X.; de Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.;
Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356. [CrossRef]
8. Prior, I.A.; Hood, F.E.; Hartley, J.L. The Frequency of Ras Mutations in Cancer. Cancer Res. 2020, 80, 2969–2974. [CrossRef]
Cancers 2024, 16, 2796 17 of 25
9. Neumann, J.; Zeindl-Eberhart, E.; Kirchner, T.; Jung, A. Frequency and type of KRAS mutations in routine diagnostic analysis of
metastatic colorectal cancer. Pathol. Res. Pr. 2009, 205, 858–862. [CrossRef]
10. Zhu, C.; Guan, X.; Zhang, X.; Luan, X.; Song, Z.; Cheng, X.; Zhang, W.; Qin, J.J. Targeting KRAS mutant cancers: From druggable
therapy to drug resistance. Mol. Cancer 2022, 21, 159. [CrossRef]
11. Smith, J.C.; Brooks, L.; Hoff, P.M.; McWalter, G.; Dearden, S.; Morgan, S.R.; Wilson, D.; Robertson, J.D.; Jürgensmeier, J.M. KRAS
mutations are associated with inferior clinical outcome in patients with metastatic colorectal cancer, but are not predictive for
benefit with cediranib. Eur. J. Cancer 2013, 49, 2424–2432. [CrossRef]
12. Aljehani, M.A.; Bien, J.; Lee, J.S.H.; Fisher, G.A.; Lin, A.Y. KRAS Sequence Variation as Prognostic Marker in Patients With Young-
vs Late-Onset Colorectal Cancer. JAMA Netw. Open 2023, 6, e2345801. [CrossRef] [PubMed]
13. Ottaiano, A.; Sabbatino, F.; Perri, F.; Cascella, M.; Sirica, R.; Patrone, R.; Capuozzo, M.; Savarese, G.; Ianniello, M.; Petrillo, N.; et al.
p.G12C Mutation in Metastatic Colorectal Cancer: Prognostic Implications and Advancements in Targeted Therapies. Cancers
2023, 15, 3579. [CrossRef] [PubMed]
14. Van Cutsem, E.; Köhne, C.H.; Láng, I.; Folprecht, G.; Nowacki, M.P.; Cascinu, S.; Shchepotin, I.; Maurel, J.; Cunningham, D.;
Tejpar, S.; et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer:
Updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J. Clin. Oncol. 2011, 29, 2011–2019.
[CrossRef] [PubMed]
15. Douillard, J.Y.; Oliner, K.S.; Siena, S.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.;
et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N. Engl. J. Med. 2013, 369, 1023–1034. [CrossRef]
[PubMed]
16. National Comprehensive Cancer Network. Colon Cancer (Version 3.2024). Available online: https://www.nccn.org/
professionals/physician_gls/pdf/colon.pdf (accessed on 16 June 2024).
17. Diaz, L.A.; Williams, R.T.; Wu, J.; Kinde, I.; Hecht, J.R.; Berlin, J.; Allen, B.; Bozic, I.; Reiter, J.G.; Nowak, M.A.; et al. The molecular
evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012, 486, 537–540. [CrossRef] [PubMed]
18. Misale, S.; Yaeger, R.; Hobor, S.; Scala, E.; Janakiraman, M.; Liska, D.; Valtorta, E.; Schiavo, R.; Buscarino, M.; Siravegna, G.; et al.
Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012, 486, 532–536.
[CrossRef]
19. Watanabe, J.; Muro, K.; Shitara, K.; Yamazaki, K.; Shiozawa, M.; Ohori, H.; Takashima, A.; Yokota, M.; Makiyama, A.; Akazawa,
N.; et al. Panitumumab vs Bevacizumab Added to Standard First-line Chemotherapy and Overall Survival Among Patients With
RAS Wild-type, Left-Sided Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 2023, 329, 1271–1282. [CrossRef]
20. Stintzing, S.; Modest, D.P.; Rossius, L.; Lerch, M.M.; von Weikersthal, L.F.; Decker, T.; Kiani, A.; Vehling-Kaiser, U.; Al-Batran,
S.E.; Heintges, T.; et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab for metastatic colorectal cancer (FIRE-3): A
post-hoc analysis of tumour dynamics in the final RAS wild-type subgroup of this randomised open-label phase 3 trial. Lancet
Oncol. 2016, 17, 1426–1434. [CrossRef]
21. Xue, J.Y.; Zhao, Y.; Aronowitz, J.; Mai, T.T.; Vides, A.; Qeriqi, B.; Kim, D.; Li, C.; de Stanchina, E.; Mazutis, L.; et al. Rapid
non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature 2020, 577, 421–425. [CrossRef]
22. Yaeger, R.; Weiss, J.; Pelster, M.S.; Spira, A.I.; Barve, M.; Ou, S.I.; Leal, T.A.; Bekaii-Saab, T.S.; Paweletz, C.P.; Heavey, G.A.; et al.
Adagrasib with or without Cetuximab in Colorectal Cancer with Mutated. N. Engl. J. Med. 2023, 388, 44–54. [CrossRef] [PubMed]
23. Fakih, M.G.; Salvatore, L.; Esaki, T.; Modest, D.P.; Lopez-Bravo, D.P.; Taieb, J.; Karamouzis, M.V.; Ruiz-Garcia, E.; Kim, T.W.;
Kuboki, Y.; et al. Sotorasib plus Panitumumab in Refractory Colorectal Cancer with Mutated. N. Engl. J. Med. 2023, 389, 2125–2139.
[CrossRef] [PubMed]
24. Fedele, C.; Li, S.; Teng, K.W.; Foster, C.J.R.; Peng, D.; Ran, H.; Mita, P.; Geer, M.J.; Hattori, T.; Koide, A.; et al. SHP2 inhibition
diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J. Exp. Med. 2021, 218, e20201414. [CrossRef]
[PubMed]
25. Kandasamy, P.; Zlobec, I.; Nydegger, D.T.; Pujol-Giménez, J.; Bhardwaj, R.; Shirasawa, S.; Tsunoda, T.; Hediger, M.A. Oncogenic
KRAS mutations enhance amino acid uptake by colorectal cancer cells via the hippo signaling effector YAP1. Mol. Oncol. 2021, 15,
2782–2800. [CrossRef] [PubMed]
26. Mukhopadhyay, S.; Huang, H.Y.; Lin, Z.; Ranieri, M.; Li, S.; Sahu, S.; Liu, Y.; Ban, Y.; Guidry, K.; Hu, H.; et al. Genome-Wide
CRISPR Screens Identify Multiple Synthetic Lethal Targets That Enhance KRASG12C Inhibitor Efficacy. Cancer Res. 2023, 83,
4095–4111. [CrossRef] [PubMed]
27. Tajiknia, V.; El-Deiry, W.S.; Schwermann, M.; Huntington, K.; Zhou, L.; Srinivasan, P. Combination of 5-FU plus KRAS G12D
inhibitor MRTX1133 against human colorectal and pancreatic cancer cells and the affects on inhibition of pERK and immune-
stimulatory cytokine patterns in in KRAS G12D and KRAS G12V tumor cells. J. Clin. Oncol. 2023, 41, e16301. [CrossRef]
28. Lu, D.; Chen, Y.; Jiang, M.; Wang, J.; Li, Y.; Ma, K.; Sun, W.; Zheng, X.; Qi, J.; Jin, W.; et al. KRAS G12V neoantigen specific T cell
receptor for adoptive T cell therapy against tumors. Nat. Commun. 2023, 14, 6389. [CrossRef] [PubMed]
29. Singhal, A.; Li, B.T.; O’Reilly, E.M. Targeting KRAS in cancer. Nat. Med. 2024, 30, 969–983. [CrossRef] [PubMed]
30. Lavoie, H.; Therrien, M. Regulation of RAF protein kinases in ERK signalling. Nat. Rev. Mol. Cell Biol. 2015, 16, 281–298.
[CrossRef]
Cancers 2024, 16, 2796 18 of 25
31. Cope, N.; Candelora, C.; Wong, K.; Kumar, S.; Nan, H.; Grasso, M.; Novak, B.; Li, Y.; Marmorstein, R.; Wang, Z. Mechanism of
BRAF Activation through Biochemical Characterization of the Recombinant Full-Length Protein. Chembiochem 2018, 19, 1988–1997.
[CrossRef]
32. Horbinski, C. To BRAF or not to BRAF: Is that even a question anymore? J. Neuropathol. Exp. Neurol. 2013, 72, 2–7. [CrossRef]
[PubMed]
33. Cantwell-Dorris, E.R.; O’Leary, J.J.; Sheils, O.M. BRAFV600E: Implications for carcinogenesis and molecular therapy. Mol. Cancer
Ther. 2011, 10, 385–394. [CrossRef] [PubMed]
34. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al.
Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [CrossRef] [PubMed]
35. Potocki, P.M.; Wojcik, P.; Chmura, L.; Goc, B.; Fedewicz, M.; Bielanska, Z.; Swadzba, J.; Konopka, K.; Kwinta, L.; Wysocki, P.J.
Clinical Characterization of Targetable Mutations (BRAF V600E and KRAS G12C) in Advanced Colorectal Cancer-A Nation-Wide
Study. Int. J. Mol. Sci. 2023, 24, 9073. [CrossRef] [PubMed]
36. Seligmann, J.F.; Fisher, D.; Smith, C.G.; Richman, S.D.; Elliott, F.; Brown, S.; Adams, R.; Maughan, T.; Quirke, P.; Cheadle, J.; et al.
Investigating the poor outcomes of BRAF-mutant advanced colorectal cancer: Analysis from 2530 patients in randomised clinical
trials. Ann. Oncol. 2017, 28, 562–568. [CrossRef] [PubMed]
37. Tol, J.; Nagtegaal, I.D.; Punt, C.J. BRAF mutation in metastatic colorectal cancer. N. Engl. J. Med. 2009, 361, 98–99. [CrossRef]
[PubMed]
38. Price, T.J.; Hardingham, J.E.; Lee, C.K.; Weickhardt, A.; Townsend, A.R.; Wrin, J.W.; Chua, A.; Shivasami, A.; Cummins, M.M.;
Murone, C.; et al. Impact of KRAS and BRAF Gene Mutation Status on Outcomes From the Phase III AGITG MAX Trial of
Capecitabine Alone or in Combination With Bevacizumab and Mitomycin in Advanced Colorectal Cancer. J. Clin. Oncol. 2011, 29,
2675–2682. [CrossRef] [PubMed]
39. Sanz-Garcia, E.; Argiles, G.; Elez, E.; Tabernero, J. BRAF mutant colorectal cancer: Prognosis, treatment, and new perspectives.
Ann. Oncol. 2017, 28, 2648–2657. [CrossRef] [PubMed]
40. Souglakos, J.; Philips, J.; Wang, R.; Marwah, S.; Silver, M.; Tzardi, M.; Silver, J.; Ogino, S.; Hooshmand, S.; Kwak, E.; et al.
Prognostic and predictive value of common mutations for treatment response and survival in patients with metastatic colorectal
cancer. Br. J. Cancer 2009, 101, 465–472. [CrossRef]
41. Jones, J.C.; Renfro, L.A.; Al-Shamsi, H.O.; Schrock, A.B.; Rankin, A.; Zhang, B.Y.; Kasi, P.M.; Voss, J.S.; Leal, A.D.; Sun, J.; et al.
(Non-V600) BRAF Mutations Define a Clinically Distinct Molecular Subtype of Metastatic Colorectal Cancer. J. Clin. Oncol. 2017,
35, 2624–2630. [CrossRef]
42. Seymour, M.T.; Brown, S.R.; Middleton, G.; Maughan, T.; Richman, S.; Gwyther, S.; Lowe, C.; Seligmann, J.F.; Wadsley, J.; Maisey,
N.; et al. Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced
colorectal cancer (PICCOLO): A prospectively stratified randomised trial. Lancet Oncol. 2013, 14, 749–759. [CrossRef] [PubMed]
43. Pietrantonio, F.; Petrelli, F.; Coinu, A.; Di Bartolomeo, M.; Borgonovo, K.; Maggi, C.; Cabiddu, M.; Iacovelli, R.; Bossi, I.; Lonati, V.;
et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: A
meta-analysis. Eur. J. Cancer 2015, 51, 587–594. [CrossRef] [PubMed]
44. Rowland, A.; Dias, M.M.; Wiese, M.D.; Kichenadasse, G.; McKinnon, R.A.; Karapetis, C.S.; Sorich, M.J. Meta-analysis of BRAF
mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic
colorectal cancer. Br. J. Cancer 2015, 112, 1888–1894. [CrossRef] [PubMed]
45. Shinozaki, E.; Yoshino, T.; Yamazaki, K.; Muro, K.; Yamaguchi, K.; Nishina, T.; Yuki, S.; Shitara, K.; Bando, H.; Mimaki, S.; et al.
Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment
in patients with pretreated metastatic colorectal cancer: The Biomarker Research for anti-EGFR monoclonal Antibodies by
Comprehensive Cancer genomics (BREAC) study. Br. J. Cancer 2017, 117, 1450–1458. [CrossRef] [PubMed]
46. Yaeger, R.; Kotani, D.; Mondaca, S.; Parikh, A.R.; Bando, H.; Van Seventer, E.E.; Taniguchi, H.; Zhao, H.; Thant, C.N.; de Stanchina,
E.; et al. Response to Anti-EGFR Therapy in Patients with BRAF non-V600-Mutant Metastatic Colorectal Cancer. Clin. Cancer Res.
2019, 25, 7089–7097. [CrossRef] [PubMed]
47. Hanrahan, A.J.; Solit, D.B. BRAF Mutations: The Discovery of Allele- and Lineage-Specific Differences. Cancer Res. 2022, 82,
12–14. [CrossRef] [PubMed]
48. Yao, Z.; Torres, N.M.; Tao, A.; Gao, Y.; Luo, L.; Li, Q.; de Stanchina, E.; Abdel-Wahab, O.; Solit, D.B.; Poulikakos, P.I.; et al. BRAF
Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition.
Cancer Cell 2015, 28, 370–383. [CrossRef] [PubMed]
49. Middleton, G.; Yang, Y.; Campbell, C.D.; Andre, T.; Atreya, C.E.; Schellens, J.H.M.; Yoshino, T.; Bendell, J.C.; Hollebecque, A.;
McRee, A.J.; et al. BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with
Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer. Clin. Cancer Res. 2020, 26, 2466–2476. [CrossRef]
50. Kopetz, S.; Grothey, A.; Yaeger, R.; Van Cutsem, E.; Desai, J.; Yoshino, T.; Wasan, H.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al.
Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. N. Engl. J. Med. 2019, 381, 1632–1643.
[CrossRef]
51. Tabernero, J.; Grothey, A.; Van Cutsem, E.; Yaeger, R.; Wasan, H.; Yoshino, T.; Desai, J.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al.
Encorafenib Plus Cetuximab as a New Standard of Care for Previously Treated. J. Clin. Oncol. 2021, 39, 273–284. [CrossRef]
Cancers 2024, 16, 2796 19 of 25
52. Gandini, A.; Puglisi, S.; Pirrone, C.; Martelli, V.; Catalano, F.; Nardin, S.; Seeber, A.; Puccini, A.; Sciallero, S. The role of
immunotherapy in microsatellites stable metastatic colorectal cancer: State of the art and future perspectives. Front. Oncol. 2023,
13, 1161048. [CrossRef] [PubMed]
53. Morris, V.K.; Parseghian, C.M.; Escano, M.; Johnson, B.; Raghav, K.P.S.; Dasari, A.; Huey, R.; Overman, M.J.; Willis, J.; Lee, M.S.;
et al. Phase I/II trial of encorafenib, cetuximab, and nivolumab in patients with microsatellite stable, BRAFV600E metastatic
colorectal cancer. J. Clin. Oncol. 2022, 40, 12. [CrossRef]
54. Valtorta, E.; Martino, C.; Sartore-Bianchi, A.; Penaullt-Llorca, F.; Viale, G.; Risio, M.; Rugge, M.; Grigioni, W.; Bencardino, K.;
Lonardi, S.; et al. Assessment of a HER2 scoring system for colorectal cancer: Results from a validation study. Mod. Pathol. 2015,
28, 1481–1491. [CrossRef] [PubMed]
55. Sartore-Bianchi, A.; Amatu, A.; Porcu, L.; Ghezzi, S.; Lonardi, S.; Leone, F.; Bergamo, F.; Fenocchio, E.; Martinelli, E.; Borelli, B.;
et al. HER2 Positivity Predicts Unresponsiveness to EGFR-Targeted Treatment in Metastatic Colorectal Cancer. Oncologist 2019,
24, 1395–1402. [CrossRef] [PubMed]
56. Sartore-Bianchi, A.; Trusolino, L.; Martino, C.; Bencardino, K.; Lonardi, S.; Bergamo, F.; Zagonel, V.; Leone, F.; Depetris, I.;
Martinelli, E.; et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type,
HER2-positive metastatic colorectal cancer (HERACLES): A proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol.
2016, 17, 738–746. [CrossRef] [PubMed]
57. Raghav, K.; Loree, J.M.; Morris, J.S.; Overman, M.J.; Yu, R.; Meric-Bernstam, F.; Menter, D.; Korphaisarn, K.; Kee, B.; Muranyi, A.;
et al. Validation of HER2 Amplification as a Predictive Biomarker for Anti–Epidermal Growth Factor Receptor Antibody Therapy
in Metastatic Colorectal Cancer. JCO Precis. Oncol. 2019, 3, 1–13. [CrossRef] [PubMed]
58. Martin, V.; Landi, L.; Molinari, F.; Fountzilas, G.; Geva, R.; Riva, A.; Saletti, P.; De Dosso, S.; Spitale, A.; Tejpar, S.; et al. HER2 gene
copy number status may influence clinical efficacy to anti-EGFR monoclonal antibodies in metastatic colorectal cancer patients.
Br. J. Cancer 2013, 108, 668–675. [CrossRef]
59. Battaglin, F.; Ou, F.S.; Qu, X.; Hochster, H.S.; Niedzwiecki, D.; Goldberg, R.M.; Mayer, R.J.; Ashouri, K.; Zemla, T.J.; Blanke,
C.D.; et al. Gene Expression Levels Are Predictive and Prognostic in Patients With Metastatic Colorectal Cancer Enrolled in
CALGB/SWOG 80405. J. Clin. Oncol. 2024, 42, 1890–1902. [CrossRef] [PubMed]
60. Hashimoto, T.; Nakamura, Y.; Yang, Y.; Battaglin, F.; Innocenti, F.; Venook, A.P.; Makiyama, A.; Boku, S.; Machida, N.; Takahashi,
N.; et al. Prognostic impact of HER family expressions for metastatic colorectal cancer (mCRC): SCRUM-Japan MONSTAR-
SCREEN-2 and CALGB/SWOG 80405 trial (Alliance) international collaboration. J. Clin. Oncol. 2024, 42, 3548. [CrossRef]
61. Wu, S.W.; Ma, C.C.; Li, W.H. Does overexpression of HER-2 correlate with clinicopathological characteristics and prognosis in
colorectal cancer? Evidence from a meta-analysis. Diagn. Pathol. 2015, 10, 144. [CrossRef]
62. Hainsworth, J.D.; Meric-Bernstam, F.; Swanton, C.; Hurwitz, H.; Spigel, D.R.; Sweeney, C.; Burris, H.; Bose, R.; Yoo, B.; Stein, A.;
et al. Targeted Therapy for Advanced Solid Tumors on the Basis of Molecular Profiles: Results From MyPathway, an Open-Label,
Phase IIa Multiple Basket Study. J. Clin. Oncol. 2018, 36, 536–542. [CrossRef] [PubMed]
63. Strickler, J.H.; Cercek, A.; Siena, S.; André, T.; Ng, K.; Van Cutsem, E.; Wu, C.; Paulson, A.S.; Hubbard, J.M.; Coveler, A.L.; et al.
Tucatinib plus trastuzumab for chemotherapy-refractory, HER2-positive, RAS wild-type unresectable or metastatic colorectal
cancer (MOUNTAINEER): A multicentre, open-label, phase 2 study. Lancet Oncol. 2023, 24, 496–508. [CrossRef] [PubMed]
64. Yoshino, T.; Di Bartolomeo, M.; Raghav, K.; Masuishi, T.; Loupakis, F.; Kawakami, H.; Yamaguchi, K.; Nishina, T.; Wainberg,
Z.; Elez, E.; et al. Final results of DESTINY-CRC01 investigating trastuzumab deruxtecan in patients with HER2-expressing
metastatic colorectal cancer. Nat. Commun. 2023, 14, 3332. [CrossRef]
65. Bonneville, R.; Krook, M.A.; Kautto, E.A.; Miya, J.; Wing, M.R.; Chen, H.Z.; Reeser, J.W.; Yu, L.; Roychowdhury, S. Landscape of
Microsatellite Instability Across 39 Cancer Types. JCO Precis. Oncol. 2017, 1, 1–15. [CrossRef] [PubMed]
66. Ligtenberg, M.J.; Kuiper, R.P.; Geurts van Kessel, A.; Hoogerbrugge, N. EPCAM deletion carriers constitute a unique subgroup of
Lynch syndrome patients. Fam. Cancer 2013, 12, 169–174. [CrossRef] [PubMed]
67. Vilar, E.; Gruber, S.B. Microsatellite instability in colorectal cancer-the stable evidence. Nat. Rev. Clin. Oncol. 2010, 7, 153–162.
[CrossRef] [PubMed]
68. Peltomäki, P.; Nyström, M.; Mecklin, J.P.; Seppälä, T.T. Lynch Syndrome Genetics and Clinical Implications. Gastroenterology 2023,
164, 783–799. [CrossRef]
69. Le, D.T.; Diaz, L.A.; Kim, T.W.; Van Cutsem, E.; Geva, R.; Jäger, D.; Hara, H.; Burge, M.; O’Neil, B.H.; Kavan, P.; et al.
Pembrolizumab for previously treated, microsatellite instability-high/mismatch repair-deficient advanced colorectal cancer:
Final analysis of KEYNOTE-164. Eur. J. Cancer 2023, 186, 185–195. [CrossRef] [PubMed]
70. Chen, C.; Liu, S.; Qu, R.; Li, B. Recurrent Neoantigens in Colorectal Cancer as Potential Immunotherapy Targets. Biomed. Res. Int.
2020, 2020, 2861240. [CrossRef]
71. Grasso, C.S.; Giannakis, M.; Wells, D.K.; Hamada, T.; Mu, X.J.; Quist, M.; Nowak, J.A.; Nishihara, R.; Qian, Z.R.; Inamura, K.; et al.
Genetic Mechanisms of Immune Evasion in Colorectal Cancer. Cancer Discov. 2018, 8, 730–749. [CrossRef]
72. Ros, J.; Baraibar, I.; Saoudi, N.; Rodriguez, M.; Salvà, F.; Tabernero, J.; Élez, E. Immunotherapy for Colorectal Cancer with High
Microsatellite Instability: The Ongoing Search for Biomarkers. Cancers 2023, 15, 4245. [CrossRef]
73. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al.
PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [CrossRef]
Cancers 2024, 16, 2796 20 of 25
74. Le, D.T.; Kim, T.W.; Van Cutsem, E.; Geva, R.; Jäger, D.; Hara, H.; Burge, M.; O’Neil, B.; Kavan, P.; Yoshino, T.; et al. Phase
II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability-High/Mismatch Repair-Deficient
Metastatic Colorectal Cancer: KEYNOTE-164. J. Clin. Oncol. 2020, 38, 11–19. [CrossRef]
75. Overman, M.J.; McDermott, R.; Leach, J.L.; Lonardi, S.; Lenz, H.J.; Morse, M.A.; Desai, J.; Hill, A.; Axelson, M.; Moss, R.A.;
et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer
(CheckMate 142): An open-label, multicentre, phase 2 study. Lancet Oncol. 2017, 18, 1182–1191. [CrossRef] [PubMed]
76. André, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al.
Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N. Engl. J. Med. 2020, 383, 2207–2218. [CrossRef]
[PubMed]
77. Lenz, H.J.; Van Cutsem, E.; Luisa Limon, M.; Wong, K.Y.M.; Hendlisz, A.; Aglietta, M.; García-Alfonso, P.; Neyns, B.; Luppi,
G.; Cardin, D.B.; et al. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-
Deficient Metastatic Colorectal Cancer: The Phase II CheckMate 142 Study. J. Clin. Oncol. 2022, 40, 161–170. [CrossRef]
[PubMed]
78. Heregger, R.; Huemer, F.; Steiner, M.; Gonzalez-Martinez, A.; Greil, R.; Weiss, L. Unraveling Resistance to Immunotherapy in
MSI-High Colorectal Cancer. Cancers 2023, 15, 5090. [CrossRef]
79. Kato, Y.; Tabata, K.; Kimura, T.; Yachie-Kinoshita, A.; Ozawa, Y.; Yamada, K.; Ito, J.; Tachino, S.; Hori, Y.; Matsuki, M.; et al.
Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated
macrophage and activation of the interferon pathway. PLoS ONE 2019, 14, e0212513. [CrossRef]
80. Doleschel, D.; Hoff, S.; Koletnik, S.; Rix, A.; Zopf, D.; Kiessling, F.; Lederle, W. Regorafenib enhances anti-PD1 immunotherapy
efficacy in murine colorectal cancers and their combination prevents tumor regrowth. J. Exp. Clin. Cancer Res. 2021, 40, 288.
[CrossRef]
81. Bendell, J.C.; Powderly, J.D.; Lieu, C.H.; Eckhardt, S.G.; Hurwitz, H.; Hochster, H.S.; Murphy, J.E.; Funke, R.P.; Rossi, C.; Wallin, J.;
et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) and/or FOLFOX in patients (pts)
with metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2015, 33, 704. [CrossRef]
82. Schrock, A.B.; Ouyang, C.; Sandhu, J.; Sokol, E.; Jin, D.; Ross, J.S.; Miller, V.A.; Lim, D.; Amanam, I.; Chao, J.; et al. Tumor
mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann.
Oncol. 2019, 30, 1096–1103. [CrossRef]
83. Doig, K.D.; Fellowes, A.; Scott, P.; Fox, S.B. Tumour mutational burden: An overview for pathologists. Pathology 2022, 54, 249–253.
[CrossRef] [PubMed]
84. Wang, J.; Song, J.; Liu, Z.; Zhang, T.; Liu, Y. High tumor mutation burden indicates better prognosis in colorectal cancer patients
with KRAS mutations. Front. Oncol. 2022, 12, 1015308. [CrossRef] [PubMed]
85. Xiao, J.; Li, W.; Huang, Y.; Huang, M.; Li, S.; Zhai, X.; Zhao, J.; Gao, C.; Xie, W.; Qin, H.; et al. A next-generation sequencing-based
strategy combining microsatellite instability and tumor mutation burden for comprehensive molecular diagnosis of advanced
colorectal cancer. BMC Cancer 2021, 21, 282. [CrossRef] [PubMed]
86. Chan, T.A.; Yarchoan, M.; Jaffee, E.; Swanton, C.; Quezada, S.A.; Stenzinger, A.; Peters, S. Development of tumor mutation burden
as an immunotherapy biomarker: Utility for the oncology clinic. Ann. Oncol. 2019, 30, 44–56. [CrossRef] [PubMed]
87. Zheng, M. Tumor mutation burden for predicting immune checkpoint blockade response: The more, the better. J. Immunother.
Cancer 2022, 10, e003087. [CrossRef] [PubMed]
88. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15,
731–747. [CrossRef] [PubMed]
89. Manea, C.A.; Badiu, D.C.; Ploscaru, I.C.; Zgura, A.; Bacinschi, X.; Smarandache, C.G.; Serban, D.; Popescu, C.G.; Grigorean, V.T.;
Botnarciuc, V. A review of NTRK fusions in cancer. Ann. Med. Surg. 2022, 79, 103893. [CrossRef] [PubMed]
90. Wang, H.; Li, Z.W.; Ou, Q.; Wu, X.; Nagasaka, M.; Shao, Y.; Ou, S.I.; Yang, Y. NTRK fusion positive colorectal cancer is a unique
subset of CRC with high TMB and microsatellite instability. Cancer Med. 2022, 11, 2541–2549. [CrossRef] [PubMed]
91. Hibar, D.P.; Demetri, G.D.; Peters, S.; Davies, J.; Humblet, O.; Maund, S.L.; Perez, L. Real-world survival outcomes in patients
with locally advanced or metastatic NTRK fusion-positive solid tumors receiving standard-of-care therapies other than targeted
TRK inhibitors. PLoS ONE 2022, 17, e0270571. [CrossRef]
92. Boussiotis, V.A. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N. Engl. J. Med. 2016, 375, 1767–1778.
[CrossRef] [PubMed]
93. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [PubMed]
94. Wang, X.; Teng, F.; Kong, L.; Yu, J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets
Ther. 2016, 9, 5023–5039. [CrossRef] [PubMed]
95. Aguilar, E.J.; Ricciuti, B.; Gainor, J.F.; Kehl, K.L.; Kravets, S.; Dahlberg, S.; Nishino, M.; Sholl, L.M.; Adeni, A.; Subegdjo, S.; et al.
Outcomes to first-line pembrolizumab in patients with non-small-cell lung cancer and very high PD-L1 expression. Ann. Oncol.
2019, 30, 1653–1659. [CrossRef] [PubMed]
96. Xin, H.; Zhou, C.; Wang, G.; Liu, Y.; Zhang, J.; Li, B.; Su, M.; Li, Z. Heterogeneity of PD-L1 expression and CD8 lymphocyte
infiltration in metastatic colorectal cancer and their prognostic significance. Heliyon 2023, 9, e13048. [CrossRef] [PubMed]
Cancers 2024, 16, 2796 21 of 25
97. André, T.; Overman, M.; Lonardi, S.; Aglietta, M.; McDermott, R.; Wong, K.Y.M.; Morse, M.; Hendlisz, A.; Moss, R.A.; Ledeine,
J.M.; et al. Analysis of tumor PD-L1 expression and biomarkers in relation to clinical activity in patients (pts) with deficient DNA
mismatch repair (dMMR)/high microsatellite instability (MSI-H) metastatic colorectal cancer (mCRC) treated with nivolumab
(NIVO) + ipilimumab (IPI): CheckMate 142. Ann. Oncol. 2017, 28, v163. [CrossRef]
98. Wang, S.; Yuan, B.; Wang, Y.; Li, M.; Liu, X.; Cao, J.; Li, C.; Hu, J. Clinicopathological and prognostic significance of PD-
L1 expression in colorectal cancer: A meta-analysis. Int. J. Color. Dis. 2021, 36, 117–130. [CrossRef]
99. Yang, L.; Xue, R.; Pan, C. Prognostic and clinicopathological value of PD-L1 in colorectal cancer: A systematic review and
meta-analysis. Onco Targets Ther. 2019, 12, 3671–3682. [CrossRef]
100. Li, Y.; Liang, L.; Dai, W.; Cai, G.; Xu, Y.; Li, X.; Li, Q.; Cai, S. Prognostic impact of programed cell death-1 (PD-1) and PD-ligand 1
(PD-L1) expression in cancer cells and tumor infiltrating lymphocytes in colorectal cancer. Mol. Cancer 2016, 15, 55. [CrossRef]
101. Lee, K.S.; Kwak, Y.; Ahn, S.; Shin, E.; Oh, H.K.; Kim, D.W.; Kang, S.B.; Choe, G.; Kim, W.H.; Lee, H.S. Prognostic implication
of CD274 (PD-L1) protein expression in tumor-infiltrating immune cells for microsatellite unstable and stable colorectal cancer.
Cancer Immunol. Immunother. 2017, 66, 927–939. [CrossRef]
102. Peng, Y.; Wang, Y.; Zhou, C.; Mei, W.; Zeng, C. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making
Headway? Front. Oncol. 2022, 12, 819128. [CrossRef] [PubMed]
103. Mao, C.; Yang, Z.Y.; Hu, X.F.; Chen, Q.; Tang, J.L. PIK3CA exon 20 mutations as a potential biomarker for resistance to anti-EGFR
monoclonal antibodies in KRAS wild-type metastatic colorectal cancer: A systematic review and meta-analysis. Ann. Oncol. 2012,
23, 1518–1525. [CrossRef] [PubMed]
104. Lu, X.; Li, Y.; Zhang, X.; Shi, J.; Feng, H.; Yu, Z.; Gao, Y. Prognostic and predictive biomarkers for anti-EGFR monoclonal antibody
therapy in RAS wild-type metastatic colorectal cancer: A systematic review and meta-analysis. BMC Cancer 2023, 23, 1117.
[CrossRef] [PubMed]
105. Castellano, E.; Downward, J. RAS Interaction with PI3K: More Than Just Another Effector Pathway. Genes. Cancer 2011, 2, 261–274.
[CrossRef] [PubMed]
106. Tan, E.S.; Fan, W.; Knepper, T.C.; Schell, M.J.; Sahin, I.H.; Fleming, J.B.; Xie, H. Prognostic and Predictive Value of PIK3CA
Mutations in Metastatic Colorectal Cancer. Target. Oncol. 2022, 17, 483–492. [CrossRef]
107. Goïta, A.A.; Guenot, D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers 2022, 14,
1810. [CrossRef] [PubMed]
108. Ottaiano, A.; di Palma, A.; Napolitano, M.; Pisano, C.; Pignata, S.; Tatangelo, F.; Botti, G.; Acquaviva, A.M.; Castello, G.; Ascierto,
P.A.; et al. Inhibitory effects of anti-CXCR4 antibodies on human colon cancer cells. Cancer Immunol. Immunother. 2005, 54,
781–791. [CrossRef] [PubMed]
109. Zeelenberg, I.S.; Ruuls-Van Stalle, L.; Roos, E. The chemokine receptor CXCR4 is required for outgrowth of colon carcinoma
micrometastases. Cancer Res. 2003, 63, 3833–3839.
110. Biasci, D.; Smoragiewicz, M.; Connell, C.M.; Wang, Z.; Gao, Y.; Thaventhiran, J.E.D.; Basu, B.; Magiera, L.; Johnson, T.I.; Bax, L.;
et al. CXCR4 inhibition in human pancreatic and colorectal cancers induces an integrated immune response. Proc. Natl. Acad. Sci.
USA 2020, 117, 28960–28970. [CrossRef]
111. Kim, J.; Mori, T.; Chen, S.L.; Amersi, F.F.; Martinez, S.R.; Kuo, C.; Turner, R.R.; Ye, X.; Bilchik, A.J.; Morton, D.L.; et al. Chemokine
receptor CXCR4 expression in patients with melanoma and colorectal cancer liver metastases and the association with disease
outcome. Ann. Surg. 2006, 244, 113–120. [CrossRef]
112. Kim, J.; Takeuchi, H.; Lam, S.T.; Turner, R.R.; Wang, H.J.; Kuo, C.; Foshag, L.; Bilchik, A.J.; Hoon, D.S. Chemokine receptor
CXCR4 expression in colorectal cancer patients increases the risk for recurrence and for poor survival. J. Clin. Oncol. 2005, 23,
2744–2753. [CrossRef] [PubMed]
113. Yopp, A.C.; Shia, J.; Butte, J.M.; Allen, P.J.; Fong, Y.; Jarnagin, W.R.; DeMatteo, R.P.; D’Angelica, M.I. CXCR4 expression predicts
patient outcome and recurrence patterns after hepatic resection for colorectal liver metastases. Ann. Surg. Oncol. 2012, 19 (Suppl.
S3), S339–S346. [CrossRef] [PubMed]
114. Huang, W.S.; Hsieh, M.C.; Huang, C.Y.; Kuo, Y.H.; Tung, S.Y.; Shen, C.H.; Hsieh, Y.Y.; Teng, C.C.; Lee, K.F.; Chen, T.C.; et al. The
Association of CXC Receptor 4 Mediated Signaling Pathway with Oxaliplatin-Resistant Human Colorectal Cancer Cells. PLoS
ONE 2016, 11, e0159927. [CrossRef] [PubMed]
115. Aldinucci, D.; Borghese, C.; Casagrande, N. The CCL5/CCR5 Axis in Cancer Progression. Cancers 2020, 12, 1765. [CrossRef]
[PubMed]
116. Schlecker, E.; Stojanovic, A.; Eisen, C.; Quack, C.; Falk, C.S.; Umansky, V.; Cerwenka, A. Tumor-infiltrating monocytic myeloid-
derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J. Immunol. 2012,
189, 5602–5611. [CrossRef] [PubMed]
117. Battaglin, F.; Baca, Y.; Millstein, J.; Yang, Y.; Xiu, J.; Arai, H.; Wang, J.; Ou, F.S.; Innocenti, F.; Mumenthaler, S.M.; et al. CCR5 and
CCL5 gene expression in colorectal cancer: Comprehensive profiling and clinical value. J. Immunother. Cancer 2024, 12, e007939.
[CrossRef] [PubMed]
118. Suenaga, M.; Cao, S.; Zhang, W.; Yang, D.; Ning, Y.; Okazaki, S.; Berger, M.D.; Miyamoto, Y.; Schirripa, M.; Soni, S.; et al. Genetic
variants in CCL5 and CCR5 genes and serum VEGF-A levels predict efficacy of bevacizumab in metastatic colorectal cancer
patients. Int. J. Cancer 2019, 144, 2567–2577. [CrossRef]
Cancers 2024, 16, 2796 22 of 25
119. Suenaga, M.; Schirripa, M.; Cao, S.; Zhang, W.; Yang, D.; Ning, Y.; Cremolini, C.; Antoniotti, C.; Borelli, B.; Mashima, T.; et al. Gene
Polymorphisms in the CCL5/CCR5 Pathway as a Genetic Biomarker for Outcome and Hand-Foot Skin Reaction in Metastatic
Colorectal Cancer Patients Treated With Regorafenib. Clin. Color. Cancer 2018, 17, e395–e414. [CrossRef] [PubMed]
120. Suenaga, M.; Zhang, W.U.; Mashima, T.; Schirripa, M.; Cao, S.; Okazaki, S.; Berger, M.D.; Miyamoto, Y.; Barzi, A.; Yamaguchi,
T.; et al. Potential Molecular Cross Talk Among CCR5 Pathway Predicts Regorafenib Responsiveness in Metastatic Colorectal
Cancer Patients. Cancer Genom. Proteom. 2021, 18, 317–324. [CrossRef]
121. Suarez-Carmona, M.; Chaorentong, P.; Kather, J.N.; Rothenheber, R.; Ahmed, A.; Berthel, A.; Heinzelmann, A.; Moraleda, R.;
Valous, N.A.; Kosaloglu, Z.; et al. CCR5 status and metastatic progression in colorectal cancer. Oncoimmunology 2019, 8, e1626193.
[CrossRef]
122. Nygaard, V.; Ree, A.H.; Dagenborg, V.J.; Børresen-Dale, A.L.; Edwin, B.; Fretland, Å.A.; Grzyb, K.; Haugen, M.H.; Mælandsmo,
G.M.; Flatmark, K. A PRRX1 Signature Identifies TIM-3 and VISTA as Potential Immune Checkpoint Targets in a Subgroup of
Microsatellite Stable Colorectal Cancer Liver Metastases. Cancer Res. Commun. 2023, 3, 235–244. [CrossRef]
123. Sauer, N.; Szlasa, W.; Jonderko, L.; Oślizło, M.; Kunachowicz, D.; Kulbacka, J.; Karłowicz-Bodalska, K. LAG-3 as a Potent Target
for Novel Anticancer Therapies of a Wide Range of Tumors. Int. J. Mol. Sci. 2022, 23, 9958. [CrossRef]
124. Katagata, M.; Okayama, H.; Nakajima, S.; Saito, K.; Sato, T.; Sakuma, M.; Fukai, S.; Endo, E.; Sakamoto, W.; Saito, M.; et al.
TIM-3 Expression and M2 Polarization of Macrophages in the TGFβ-Activated Tumor Microenvironment in Colorectal Cancer.
Cancers 2023, 15, 4943. [CrossRef]
125. Ma, Q.; Liu, J.; Wu, G.; Teng, M.; Wang, S.; Cui, M.; Li, Y. Co-expression of LAG3 and TIM3 identifies a potent Treg population
that suppresses macrophage functions in colorectal cancer patients. Clin. Exp. Pharmacol. Physiol. 2018, 45, 1002–1009. [CrossRef]
[PubMed]
126. Kuai, W.; Xu, X.; Yan, J.; Zhao, W.; Li, Y.; Wang, B.; Yuan, N.; Li, Z.; Jia, Y. Prognostic Impact of PD-1 and Tim-3 Expression in
Tumor Tissue in Stage I-III Colorectal Cancer. Biomed. Res. Int. 2020, 2020, 5294043. [CrossRef]
127. Huang, Y.H.; Zhu, C.; Kondo, Y.; Anderson, A.C.; Gandhi, A.; Russell, A.; Dougan, S.K.; Petersen, B.S.; Melum, E.; Pertel, T.; et al.
CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 2015, 517, 386–390. [CrossRef] [PubMed]
128. Woo, S.R.; Turnis, M.E.; Goldberg, M.V.; Bankoti, J.; Selby, M.; Nirschl, C.J.; Bettini, M.L.; Gravano, D.M.; Vogel, P.; Liu, C.L.;
et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape.
Cancer Res. 2012, 72, 917–927. [CrossRef]
129. Garralda, E.; Sukari, A.; Lakhani, N.J.; Patnaik, A.; Lou, Y.; Im, S.A.; Golan, T.; Geva, R.; Wermke, M.; de Miguel, M.; et al. A
first-in-human study of the anti-LAG-3 antibody favezelimab plus pembrolizumab in previously treated, advanced microsatellite
stable colorectal cancer. ESMO Open 2022, 7, 100639. [CrossRef] [PubMed]
130. Wang, S.; Ma, H.; Yan, Y.; Chen, Y.; Fu, S.; Wang, J.; Wang, Y.; Chen, H.; Liu, J. cMET promotes metastasis and epithelial-
mesenchymal transition in colorectal carcinoma by repressing RKIP. J. Cell Physiol. 2021, 236, 3963–3978. [CrossRef]
131. Lee, S.J.; Lee, J.; Park, S.H.; Park, J.O.; Lim, H.Y.; Kang, W.K.; Park, Y.S.; Kim, S.T. c-MET Overexpression in Colorectal Cancer: A
Poor Prognostic Factor for Survival. Clin. Color. Cancer 2018, 17, 165–169. [CrossRef]
132. Gao, H.; Guan, M.; Sun, Z.; Bai, C. High c-Met expression is a negative prognostic marker for colorectal cancer: A meta-analysis.
Tumour Biol. 2015, 36, 515–520. [CrossRef] [PubMed]
133. Bardelli, A.; Corso, S.; Bertotti, A.; Hobor, S.; Valtorta, E.; Siravegna, G.; Sartore-Bianchi, A.; Scala, E.; Cassingena, A.; Zecchin,
D.; et al. Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer. Cancer Discov. 2013, 3,
658–673. [CrossRef] [PubMed]
134. Song, N.; Liu, S.; Zhang, J.; Liu, J.; Xu, L.; Liu, Y.; Qu, X. Cetuximab-induced MET activation acts as a novel resistance mechanism
in colon cancer cells. Int. J. Mol. Sci. 2014, 15, 5838–5851. [CrossRef]
135. Sharma, M.; Kuboki, Y.; Camidge, D.R.; Perets, R.; Sommerhalder, D.; Yamamoto, N.; Bar, J.; Parikh, A.; Li, R.; Thiele, G.M.; et al.
Dose escalation results from a first-in-human study of ABBV-400, a novel c-Met–targeting antibody-drug conjugate, in advanced
solid tumors. J. Clin. Oncol. 2023, 41, 3015. [CrossRef]
136. Tokunaga, R.; Xiu, J.; Goldberg, R.M.; Philip, P.A.; Seeber, A.; Battaglin, F.; Arai, H.; Lo, J.H.; Naseem, M.; Puccini, A.; et al. The
impact of ARID1A mutation on molecular characteristics in colorectal cancer. Eur. J. Cancer 2020, 140, 119–129. [CrossRef]
137. Johnson, R.M.; Qu, X.; Lin, C.F.; Huw, L.Y.; Venkatanarayan, A.; Sokol, E.; Ou, F.S.; Ihuegbu, N.; Zill, O.A.; Kabbarah, O.; et al.
ARID1A mutations confer intrinsic and acquired resistance to cetuximab treatment in colorectal cancer. Nat. Commun. 2022, 13,
5478. [CrossRef]
138. Chiappa, M.; Petrella, S.; Damia, G.; Broggini, M.; Guffanti, F.; Ricci, F. Present and Future Perspective on PLK1 Inhibition in
Cancer Treatment. Front. Oncol. 2022, 12, 903016. [CrossRef]
139. Yu, Z.; Deng, P.; Chen, Y.; Liu, S.; Chen, J.; Yang, Z.; Fan, X.; Wang, P.; Cai, Z.; Wang, Y.; et al. Inhibition of the PLK1-Coupled Cell
Cycle Machinery Overcomes Resistance to Oxaliplatin in Colorectal Cancer. Adv. Sci. 2021, 8, e2100759. [CrossRef] [PubMed]
140. Ahn, D.H.; Barzi, A.; Ridinger, M.; Samuëlsz, E.; Subramanian, R.A.; Croucher, P.J.P.; Smeal, T.; Kabbinavar, F.F.; Lenz, H.J.
Onvansertib in Combination with FOLFIRI and Bevacizumab in Second-Line Treatment of KRAS-Mutant Metastatic Colorectal
Cancer: A Phase Ib Clinical Study. Clin. Cancer Res. 2024, 30, 2039–2047. [CrossRef]
141. Diaz, L.A.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [CrossRef]
Cancers 2024, 16, 2796 23 of 25
142. Keller, L.; Belloum, Y.; Wikman, H.; Pantel, K. Clinical relevance of blood-based ctDNA analysis: Mutation detection and beyond.
Br. J. Cancer 2021, 124, 345–358. [CrossRef]
143. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol.
2017, 14, 531–548. [CrossRef]
144. Nakamura, Y.; Taniguchi, H.; Ikeda, M.; Bando, H.; Kato, K.; Morizane, C.; Esaki, T.; Komatsu, Y.; Kawamoto, Y.; Takahashi, N.;
et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and
GOZILA studies. Nat. Med. 2020, 26, 1859–1864. [CrossRef] [PubMed]
145. Kastrisiou, M.; Zarkavelis, G.; Pentheroudakis, G.; Magklara, A. Clinical Application of Next-Generation Sequencing as A Liquid
Biopsy Technique in Advanced Colorectal Cancer: A Trick or A Treat? Cancers 2019, 11, 1573. [CrossRef] [PubMed]
146. Tie, J.; Cohen, J.D.; Wang, Y.; Christie, M.; Simons, K.; Lee, M.; Wong, R.; Kosmider, S.; Ananda, S.; McKendrick, J.; et al.
Circulating Tumor DNA Analyses as Markers of Recurrence Risk and Benefit of Adjuvant Therapy for Stage III Colon Cancer.
JAMA Oncol. 2019, 5, 1710–1717. [CrossRef] [PubMed]
147. Taieb, J.; Taly, V.; Henriques, J.; Bourreau, C.; Mineur, L.; Bennouna, J.; Desrame, J.; Louvet, C.; Lepere, C.; Mabro, M.; et al.
Prognostic Value and Relation with Adjuvant Treatment Duration of ctDNA in Stage III Colon Cancer: A Post Hoc Analysis of
the PRODIGE-GERCOR IDEA-France Trial. Clin. Cancer Res. 2021, 27, 5638–5646. [CrossRef]
148. Henriksen, T.V.; Tarazona, N.; Frydendahl, A.; Reinert, T.; Gimeno-Valiente, F.; Carbonell-Asins, J.A.; Sharma, S.; Renner, D.;
Hafez, D.; Roda, D.; et al. Circulating Tumor DNA in Stage III Colorectal Cancer, beyond Minimal Residual Disease Detection,
toward Assessment of Adjuvant Therapy Efficacy and Clinical Behavior of Recurrences. Clin. Cancer Res. 2022, 28, 507–517.
[CrossRef]
149. Slater, S.; Bryant, A.; Chen, H.C.; Begum, R.; Rana, I.; Aresu, M.; Peckitt, C.; Zhitkov, O.; Lazaro-Alcausi, R.; Borja, V.; et al. ctDNA
guided adjuvant chemotherapy versus standard of care adjuvant chemotherapy after curative surgery in patients with high risk
stage II or stage III colorectal cancer: A multi-centre, prospective, randomised control trial (TRACC Part C). BMC Cancer 2023, 23,
257. [CrossRef]
150. Parikh, A.R.; Leshchiner, I.; Elagina, L.; Goyal, L.; Levovitz, C.; Siravegna, G.; Livitz, D.; Rhrissorrakrai, K.; Martin, E.E.; Van
Seventer, E.E.; et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal
cancers. Nat. Med. 2019, 25, 1415–1421. [CrossRef]
151. Satam, H.; Joshi, K.; Mangrolia, U.; Waghoo, S.; Zaidi, G.; Rawool, S.; Thakare, R.P.; Banday, S.; Mishra, A.K.; Das, G.; et al.
Next-Generation Sequencing Technology: Current Trends and Advancements. Biology 2023, 12, 997. [CrossRef]
152. Malla, M.; Loree, J.M.; Kasi, P.M.; Parikh, A.R. Using Circulating Tumor DNA in Colorectal Cancer: Current and Evolving
Practices. J. Clin. Oncol. 2022, 40, 2846–2857. [CrossRef] [PubMed]
153. Hong, M.; Tao, S.; Zhang, L.; Diao, L.T.; Huang, X.; Huang, S.; Xie, S.J.; Xiao, Z.D.; Zhang, H. RNA sequencing: New technologies
and applications in cancer research. J. Hematol. Oncol. 2020, 13, 166. [CrossRef]
154. Ma, Y.; Li, J.; Zhao, X.; Ji, C.; Hu, W.; Qu, F.; Sun, Y.; Zhang, X. Multi-omics cluster defines the subtypes of CRC with distinct
prognosis and tumor microenvironment. Eur. J. Med. Res. 2024, 29, 207. [CrossRef]
155. Russo, V.; Lallo, E.; Munnia, A.; Spedicato, M.; Messerini, L.; D’Aurizio, R.; Ceroni, E.G.; Brunelli, G.; Galvano, A.; Russo, A.; et al.
Artificial Intelligence Predictive Models of Response to Cytotoxic Chemotherapy Alone or Combined to Targeted Therapy for
Metastatic Colorectal Cancer Patients: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 4012. [CrossRef]
156. Na, H.Y.; Park, Y.; Nam, S.K.; Lee, K.S.; Oh, H.K.; Kim, D.W.; Kang, S.B.; Kim, W.H.; Lee, H.S. Expression of human leukocyte
antigen class I and β2-microglobulin in colorectal cancer and its prognostic impact. Cancer Sci. 2021, 112, 91–100. [CrossRef]
[PubMed]
157. Kawazu, M.; Ueno, T.; Saeki, K.; Sax, N.; Togashi, Y.; Kanaseki, T.; Chida, K.; Kishigami, F.; Sato, K.; Kojima, S.; et al. HLA Class
I Analysis Provides Insight Into the Genetic and Epigenetic Background of Immune Evasion in Colorectal Cancer With High
Microsatellite Instability. Gastroenterology 2022, 162, 799–812. [CrossRef]
158. Mittal, P.; Battaglin, F.; Yang, Y.; Millstein, J.; Stintzing, S.; Parikh, A.R.; Soni, S.; Lo, J.H.; Torres-Gonzalez, L.; Algaze, S.; et al.
Single nucleotide polymorphisms (SNPs) in MHC class I and II genes to predict outcome in patients (pts) with metastatic
colorectal cancer (mCRC): Data from FIRE-3, MAVERICC, and TRIBE trials. J. Clin. Oncol. 2023, 41, 3578. [CrossRef]
159. Gambella, A.; Scabini, S.; Zoppoli, G.; De Silvestri, A.; Pigozzi, S.; Paudice, M.; Campora, M.; Fiocca, R.; Grillo, F.; Mastracci,
L. HLA-G as a prognostic marker in stage II/III colorectal cancer: Not quite there yet. Histochem. Cell Biol. 2022, 158, 535–543.
[CrossRef] [PubMed]
160. Mittal, P.; Singh, S.; Sinha, R.; Shrivastava, A.; Singh, A.; Singh, I.K. Myeloid cell leukemia 1 (MCL-1): Structural characteristics
and application in cancer therapy. Int. J. Biol. Macromol. 2021, 187, 999–1018. [CrossRef]
161. Healy, M.E.; Boege, Y.; Hodder, M.C.; Böhm, F.; Malehmir, M.; Scherr, A.L.; Jetzer, J.; Chan, L.K.; Parrotta, R.; Jacobs, K.; et al.
MCL1 Is Required for Maintenance of Intestinal Homeostasis and Prevention of Carcinogenesis in Mice. Gastroenterology 2020,
159, 183–199. [CrossRef]
162. Fu, D.; Pfannenstiel, L.; Demelash, A.; Phoon, Y.P.; Mayell, C.; Cabrera, C.; Liu, C.; Zhao, J.; Dermawan, J.; Patil, D.; et al.
MCL1 nuclear translocation induces chemoresistance in colorectal carcinoma. Cell Death Dis. 2022, 13, 63. [CrossRef] [PubMed]
163. Mittal, P.; Battaglin, F.; Baca, Y.; Xiu, J.; Farrell, A.P.; Soni, S.; Lo, J.H.; Torres-Gonzalez, L.; Algaze, S.; Jayachandran, P.; et al.
Characterization of MCL-1 in patients with colorectal cancer (CRC): Expression, molecular profiles, and outcomes. J. Clin. Oncol.
2023, 41, 3085. [CrossRef]
Cancers 2024, 16, 2796 24 of 25
164. Lee, Y.S.; Song, S.J.; Hong, H.K.; Oh, B.Y.; Lee, W.Y.; Cho, Y.B. The FBW7-MCL-1 axis is key in M1 and M2 macrophage-related
colon cancer cell progression: Validating the immunotherapeutic value of targeting PI3Kγ. Exp. Mol. Med. 2020, 52, 815–831.
[CrossRef] [PubMed]
165. Li, H.X. The role of circadian clock genes in tumors. Onco Targets Ther. 2019, 12, 3645–3660. [CrossRef] [PubMed]
166. Lévi, F.; Dugué, P.A.; Innominato, P.; Karaboué, A.; Dispersyn, G.; Parganiha, A.; Giacchetti, S.; Moreau, T.; Focan, C.; Waterhouse,
J.; et al. Wrist actimetry circadian rhythm as a robust predictor of colorectal cancer patients survival. Chronobiol. Int. 2014, 31,
891–900. [CrossRef]
167. Stokes, K.; Nunes, M.; Trombley, C.; Flôres, D.E.F.L.; Wu, G.; Taleb, Z.; Alkhateeb, A.; Banskota, S.; Harris, C.; Love, O.P.; et al. The
Circadian Clock Gene, Bmal1, Regulates Intestinal Stem Cell Signaling and Represses Tumor Initiation. Cell Mol. Gastroenterol.
Hepatol. 2021, 12, 1847–1872.e1840. [CrossRef]
168. Lo, J.H.; Soni, S.; Smbatyan, G.; Torres-Gonzalez, L.; Mittal, P.; Yang, Y.; Battaglin, F.; Chan, P.; Pan, Y.; Algaze, S.; et al. Abstract
7221: A compound targeting the circadian clock protein CRY2 enhances therapeutic efficacy of bevacizumab in a colorectal cancer
(CRC) xenograft model. Cancer Res. 2024, 84, 7221. [CrossRef]
169. Zhang, Y.H.; Shi, W.N.; Wu, S.H.; Miao, R.R.; Sun, S.Y.; Luo, D.D.; Wan, S.B.; Guo, Z.K.; Wang, W.Y.; Yu, X.F.; et al. SphK2 confers
5-fluorouracil resistance to colorectal cancer via upregulating H3K56ac-mediated DPD expression. Oncogene 2020, 39, 5214–5227.
[CrossRef]
170. Zhang, Y.H.; Luo, D.D.; Wan, S.B.; Qu, X.J. S1PR2 inhibitors potently reverse 5-FU resistance by downregulating DPD expression
in colorectal cancer. Pharmacol. Res. 2020, 155, 104717. [CrossRef]
171. Malier, M.; Gharzeddine, K.; Laverriere, M.H.; Marsili, S.; Thomas, F.; Decaens, T.; Roth, G.; Millet, A. Hypoxia Drives
Dihydropyrimidine Dehydrogenase Expression in Macrophages and Confers Chemoresistance in Colorectal Cancer. Cancer Res.
2021, 81, 5963–5976. [CrossRef]
172. Cui, Z.; He, S.; Wen, F.; Lu, L.; Xu, L.; Wu, H.; Wu, S. Dihydropyrimidine Dehydrogenase (DPD) as a Bridge between the Immune
Microenvironment of Colon Cancers and 5-FU Resistance. Front. Biosci. Landmark Ed. 2023, 28, 80. [CrossRef] [PubMed]
173. Zahalka, A.H.; Frenette, P.S. Nerves in cancer. Nat. Rev. Cancer 2020, 20, 143–157. [CrossRef] [PubMed]
174. Jiang, S.H.; Zhang, X.X.; Hu, L.P.; Wang, X.; Li, Q.; Zhang, X.L.; Li, J.; Gu, J.R.; Zhang, Z.G. Systemic Regulation of Cancer
Development by Neuro-Endocrine-Immune Signaling Network at Multiple Levels. Front. Cell Dev. Biol. 2020, 8, 586757.
[CrossRef] [PubMed]
175. Wan, C.; Yan, X.; Hu, B.; Zhang, X. Emerging Roles of the Nervous System in Gastrointestinal Cancer Development. Cancers 2022,
14, 3722. [CrossRef] [PubMed]
176. Battaglin, F.; Jayachandran, P.; Strelez, C.; Lenz, A.; Algaze, S.; Soni, S.; Lo, J.H.; Yang, Y.; Millstein, J.; Zhang, W.; et al.
Neurotransmitter signaling: A new frontier in colorectal cancer biology and treatment. Oncogene 2022, 41, 4769–4778. [CrossRef]
177. Kuol, N.; Davidson, M.; Karakkat, J.; Filippone, R.T.; Veale, M.; Luwor, R.; Fraser, S.; Apostolopoulos, V.; Nurgali, K. Blocking
Muscarinic Receptor 3 Attenuates Tumor Growth and Decreases Immunosuppressive and Cholinergic Markers in an Orthotopic
Mouse Model of Colorectal Cancer. Int. J. Mol. Sci. 2022, 24, 596. [CrossRef] [PubMed]
178. Ma, Y.; Chen, H.; Li, H.; Zhao, Z.; An, Q.; Shi, C. Targeting monoamine oxidase A: A strategy for inhibiting tumor growth with
both immune checkpoint inhibitors and immune modulators. Cancer Immunol. Immunother. 2024, 73, 48. [CrossRef] [PubMed]
179. Leung, E.L.; Huang, J.; Zhang, J.; Zhang, J.; Wang, M.; Zhu, Y.; Meng, Z.; Yu, H.; Neher, E.; Ma, L.; et al. Novel Anticancer Strategy
by Targeting the Gut Microbial Neurotransmitter Signaling to Overcome Immunotherapy Resistance. Antioxid. Redox Signal 2023,
38, 298–315. [CrossRef]
180. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang,
W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [CrossRef]
181. Plays, M.; Muller, S.; Rodriguez, R. Chemistry and biology of ferritin. Metallomics 2021, 13, mfab021. [CrossRef] [PubMed]
182. Zhou, B.; Liu, J.; Kang, R.; Klionsky, D.J.; Kroemer, G.; Tang, D. Ferroptosis is a type of autophagy-dependent cell death. Semin.
Cancer Biol. 2020, 66, 89–100. [CrossRef] [PubMed]
183. Liu, Y.; Guo, F.; Guo, W.; Wang, Y.; Song, W.; Fu, T. Ferroptosis-related genes are potential prognostic molecular markers for
patients with colorectal cancer. Clin. Exp. Med. 2021, 21, 467–477. [CrossRef] [PubMed]
184. Chen, Y.; Li, H. Prognostic and Predictive Models for Left- and Right- Colorectal Cancer Patients: A Bioinformatics Analysis
Based on Ferroptosis-Related Genes. Front. Oncol. 2022, 12, 833834. [CrossRef] [PubMed]
185. Yan, L.; Chen, X.; Bian, Z.; Gu, C.; Ji, H.; Chen, L.; Xu, H.; Tang, Q. A ferroptosis associated gene signature for predicting prognosis
and immune responses in patients with colorectal carcinoma. Front. Genet. 2022, 13, 971364. [CrossRef]
186. Jiang, Z.; Lim, S.O.; Yan, M.; Hsu, J.L.; Yao, J.; Wei, Y.; Chang, S.S.; Yamaguchi, H.; Lee, H.H.; Ke, B.; et al. TYRO3 induces
anti-PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J. Clin. Investig. 2021, 131, e139434.
[CrossRef] [PubMed]
187. Peng, B.; Peng, J.; Kang, F.; Zhang, W.; Peng, E.; He, Q. Ferroptosis-Related Gene MT1G as a Novel Biomarker Correlated With
Prognosis and Immune Infiltration in Colorectal Cancer. Front. Cell Dev. Biol. 2022, 10, 881447. [CrossRef]
188. Conche, C.; Finkelmeier, F.; Pesic, M.; Nicolas, A.M.; Bottger, T.W.; Kennel, K.B.; Denk, D.; Ceteci, F.; Mohs, K.; Engel, E.; et al.
Combining ferroptosis induction with MDSC blockade renders primary tumours and metastases in liver sensitive to immune
checkpoint blockade. Gut 2023, 72, 1774–1782. [CrossRef] [PubMed]
Cancers 2024, 16, 2796 25 of 25
189. Lin, X.; Chen, H.; Deng, T.; Cai, B.; Xia, Y.; Xie, L.; Wang, H.; Huang, C. Improved Immune Response for Colorectal Cancer
Therapy Triggered by Multifunctional Nanocomposites with Self-Amplifying Antitumor Ferroptosis. ACS Appl. Mater. Interfaces
2024, 16, 13481–13495. [CrossRef]
190. Gao, M.; Monian, P.; Quadri, N.; Ramasamy, R.; Jiang, X. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol. Cell 2015, 59,
298–308. [CrossRef] [PubMed]
191. Mukhopadhyay, S.; Vander Heiden, M.G.; McCormick, F. The Metabolic Landscape of RAS-Driven Cancers from biology to
therapy. Nat. Cancer 2021, 2, 271–283. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.